1
|
Ma R, Yang Z, Miao X, Hu J, Zhang T, Ma LT, Lin JY, Zhao LN. Dual-Mode Radiosensitization of Esophageal Squamous Cell Carcinoma via SOCS6-Loaded Virus-Inspired Manganese-Bismuth Bimetallic Oxide Nanoparticles. Adv Healthc Mater 2025:e2404737. [PMID: 40159874 DOI: 10.1002/adhm.202404737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Radioresistance poses a significant obstacle to controlling the recurrence of esophageal squamous cell carcinoma (ESCC) during radiotherapy. It is urgent to develop innovative radiosensitization strategies to improve the prognosis of patients with ESCC. Here, a novel dual-mode radiosensitizer: a virus-inspired hollow mesoporous manganese-bismuth bimetallic oxide nanoparticles (vHMMn-Bi) encapsulating the radiosensitizing plasmids (suppressor of cytokine signaling 6, SOCS6) is developed, designed to significantly amplify ESCC radiotherapy under hypoxic conditions. After intravenous injection, the SOCS6@vHMMn-Bi nanoparticles can be efficiently delivered to the tumor site and rapidly invade tumor cells by virus-like surface-assisted adhesion. Under X-ray irradiation, the nanoparticles exhibits a unique dual-mode sensitization effect, encompassing exogenous and endogenous mechanisms, thereby significantly augmenting the ESCC radiotherapeutic effectiveness. First, the Bi2O3 within the shell can enhance the radiosensitivity owing to its robust X-ray attenuation characteristics. Second, the SOCS6 released from the interior can inhibit both HIF-1α and JAK2/STAT3 signaling pathways, triggering ROS upregulation and intensifying radiation-mediated DNA damage inside ESCC cells. Furthermore, the shell employs MnO2 to catalyze the decomposition of endogenous H2O2 to increase oxygen generation, alleviating hypoxia within the tumor microenvironment. These nanoparticles demonstrates considerable potential as dual-mode radiosensitizers with no systemic toxicity and low immunogenicity for amplifying radiotherapeutic efficacy in ESCC.
Collapse
Affiliation(s)
- Rui Ma
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Zhi Yang
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Xia Miao
- Department of Radiation Medicine, The Faculty of Preventive Medicine, The Fourth Military Medical University, Xi'an, 710000, P. R. China
| | - Jing Hu
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Te Zhang
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| | - Li-Tian Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, P. R. China
| | - Jin-Yan Lin
- Department of Occupational and Environmental Health, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710000, P. R. China
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, P. R. China
| | - Li-Na Zhao
- Department of Radiation Oncology, Xi Jing Hospital, The Fourth Military Medical University, No. 127 West Chang Le Road, Xi'an, 710000, P. R. China
| |
Collapse
|
2
|
Mittal S, Mallia MB. Molecular imaging of tumor hypoxia: Evolution of nitroimidazole radiopharmaceuticals and insights for future development. Bioorg Chem 2023; 139:106687. [PMID: 37406518 DOI: 10.1016/j.bioorg.2023.106687] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Though growing evidence has been collected in support of the concept of dose escalation based on the molecular level images indicating hypoxic tumor sub-volumes that could be radio-resistant, validation of the concept is still a work in progress. Molecular imaging of tumor hypoxia using radiopharmaceuticals is expected to provide the required input to plan dose escalation through Image Guided Radiation Therapy (IGRT) to kill/control the radio-resistant hypoxic tumor cells. The success of the IGRT, therefore, is heavily dependent on the quality of images obtained using the radiopharmaceutical and the extent to which the image represents the true hypoxic status of the tumor in spite of the heterogeneous nature of tumor hypoxia. Available literature on radiopharmaceuticals for imaging hypoxia is highly skewed in favor of nitroimidazole as the pharmacophore given their ability to undergo oxygen dependent reduction in hypoxic cells. In this context, present review on nitroimidazole radiopharmaceuticals would be immensely helpful to the researchers to obtain a birds-eye view on what has been achieved so far and what can be tried differently to obtain a better hypoxia imaging agent. The review also covers various methods of radiolabeling that could be utilized for developing radiotracers for hypoxia targeting applications.
Collapse
Affiliation(s)
- Sweety Mittal
- Radiopharmaceuticals Division, Bhabha Atomic Research Center, Mumbai 400085, India.
| | - Madhava B Mallia
- Radiopharmaceuticals Division, Bhabha Atomic Research Center, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
3
|
Vinciguerra A, Bedarida V, Pronier C, El Zein S, Wassef M, Atallah S, Chatelet F, Molher J, Manivet P, Herman P, Adle-Biassette H, Verillaud B. Expression, Prognostic Value and Correlation with HPV Status of Hypoxia-Induced Markers in Sinonasal Squamous Cell Carcinoma. J Pers Med 2023; 13:jpm13050767. [PMID: 37240937 DOI: 10.3390/jpm13050767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: In head and neck squamous cell carcinoma, tumor hypoxia has been associated with radio/chemoresistance and poor prognosis, whereas human papillomavirus (HPV)-positive status has a positive impact on treatment response and survival outcomes. The aim of this study was to evaluate the expression and the potential prognostic value of hypoxia-induced endogenous markers in patients treated for squamous cell carcinoma of the nasal cavity and paranasal sinuses (SNSCC), and their correlation with HPV status. (2) Methods: In this monocentric study, patients treated in a curative intent for a SNSCC were screened retrospectively. Protein expression of CA-IX, GLUT-1, VEGF, VEGF-R1, and HIF-1α was determined by immunohistochemical staining, scored, and then correlated with overall survival (OS) and locoregional recurrence free survival (LRRFS). HPV status was assessed and correlated with hypoxic markers. (3) Results: 40 patients were included. A strong expression of CA-IX, GLUT-1, VEGF, and VEGF-R1 was detected in 30%, 32.5%, 50%, and 37.5% of cases, respectively. HIF-1α was detected in 27.5% of cases. High CA-IX expression was associated in univariate analysis with poor OS (p = 0.035), but there was no significant association between GLUT-1, VEGF, VEGF-R1, and HIF-1α expression, and OS/LRRFS. There was no correlation found between HPV status and hypoxia-induced endogenous markers (all p > 0.05). (4) Conclusions: This study provides data on the expression of hypoxia-induced endogenous markers in patients treated for SNSCC and underlines the potential role of CA-IX as a prognostic biomarker for SNSCC.
Collapse
Affiliation(s)
- Alessandro Vinciguerra
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
| | - Vincent Bedarida
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
- Centre de Ressources Biologiques Biobank Lariboisière (BB-0033-00064), DMU BioGem, AP-HP, 75010 Paris, France
| | - Charlotte Pronier
- Université Rennes, CHU Rennes, Virology, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) UMR_S 1085, F-35000 Rennes, France
| | - Sophie El Zein
- Pathology Department, Institut Curie, 75010 Paris, France
| | - Michel Wassef
- Pathology Department, DMU DREAM, AP-HP, Hôpital Lariboisière, 75010 Paris, France
| | - Sarah Atallah
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
- Université Paris Cité, 75010 Paris, France
| | - Florian Chatelet
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
- Université Paris Cité, 75010 Paris, France
- INSERM UMR 1153 ECSTRRA Team, 75010 Paris, France
| | - Joffrey Molher
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
| | - Philippe Manivet
- Centre de Ressources Biologiques Biobank Lariboisière (BB-0033-00064), DMU BioGem, AP-HP, 75010 Paris, France
- Université Paris Cité, 75010 Paris, France
- INSERM U1141 NeuroDiderot, 75010 Paris, France
| | - Philippe Herman
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
- Université Paris Cité, 75010 Paris, France
- INSERM U1141 NeuroDiderot, 75010 Paris, France
| | - Homa Adle-Biassette
- Pathology Department, DMU DREAM, AP-HP, Hôpital Lariboisière, 75010 Paris, France
- Université Paris Cité, 75010 Paris, France
- INSERM U1141 NeuroDiderot, 75010 Paris, France
| | - Benjamin Verillaud
- Otorhinolaryngology and Skull Base Center, AP-HP, Hôpital Lariboisière, 75010 Paris, France
- Université Paris Cité, 75010 Paris, France
- INSERM U1141 NeuroDiderot, 75010 Paris, France
| |
Collapse
|
4
|
Gouel P, Decazes P, Vera P, Gardin I, Thureau S, Bohn P. Advances in PET and MRI imaging of tumor hypoxia. Front Med (Lausanne) 2023; 10:1055062. [PMID: 36844199 PMCID: PMC9947663 DOI: 10.3389/fmed.2023.1055062] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Tumor hypoxia is a complex and evolving phenomenon both in time and space. Molecular imaging allows to approach these variations, but the tracers used have their own limitations. PET imaging has the disadvantage of low resolution and must take into account molecular biodistribution, but has the advantage of high targeting accuracy. The relationship between the signal in MRI imaging and oxygen is complex but hopefully it would lead to the detection of truly oxygen-depleted tissue. Different ways of imaging hypoxia are discussed in this review, with nuclear medicine tracers such as [18F]-FMISO, [18F]-FAZA, or [64Cu]-ATSM but also with MRI techniques such as perfusion imaging, diffusion MRI or oxygen-enhanced MRI. Hypoxia is a pejorative factor regarding aggressiveness, tumor dissemination and resistance to treatments. Therefore, having accurate tools is particularly important.
Collapse
Affiliation(s)
- Pierrick Gouel
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Pierre Decazes
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Pierre Vera
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Isabelle Gardin
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Sébastien Thureau
- QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France,Département de Radiothérapie, Centre Henri Becquerel, Rouen, France
| | - Pierre Bohn
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France,*Correspondence: Pierre Bohn,
| |
Collapse
|
5
|
Wadsworth BJ, Lee CM, Bennewith KL. Transiently hypoxic tumour cell turnover and radiation sensitivity in human tumour xenografts. Br J Cancer 2022; 126:1616-1626. [PMID: 35031765 PMCID: PMC9130130 DOI: 10.1038/s41416-021-01691-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/24/2021] [Accepted: 12/23/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Solid tumour perfusion can be unstable, creating transiently hypoxic cells that can contribute to radiation resistance. We investigated the in vivo lifetime of transiently hypoxic tumour cells and chronically hypoxic tumour cells during tumour growth and following irradiation. METHODS Hypoxic cells in SiHa and WiDr human tumour xenografts were labelled using pimonidazole and EF5, and turnover was quantified as the loss of labelled cells over time. The perfusion-modifying drug pentoxifylline was used to reoxygenate transiently hypoxic cells prior to hypoxia marker administration or irradiation. RESULTS Chronically hypoxic cells constantly turnover in SiHa and WiDr tumours, with half-lives ranging from 42-82 h and significant numbers surviving >96 h. Transiently hypoxic cells constitute 26% of the total hypoxic cells in WiDr tumours. These transiently hypoxic cells survive at least 24 h, but then rapidly turnover with a half-life of 34 h and are undetectable 72 h after labelling. Transiently hypoxic cells are radiation-resistant, although vascular dysfunction induced by 10 Gy of ionising radiation preferentially kills transiently hypoxic cells. CONCLUSIONS Transiently hypoxic tumour cells survive up to 72 h in WiDr tumours and are radiation-resistant, although transiently hypoxic cells are sensitive to vascular dysfunction induced by high doses of ionising radiation.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Che-Min Lee
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
6
|
Immune Checkpoint Inhibitors in Cancer Therapy. Curr Oncol 2022; 29:3044-3060. [PMID: 35621637 PMCID: PMC9139602 DOI: 10.3390/curroncol29050247] [Citation(s) in RCA: 566] [Impact Index Per Article: 188.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
The discovery of immune checkpoint proteins such as PD-1/PDL-1 and CTLA-4 represents a significant breakthrough in the field of cancer immunotherapy. Therefore, humanized monoclonal antibodies, targeting these immune checkpoint proteins have been utilized successfully in patients with metastatic melanoma, renal cell carcinoma, head and neck cancers and non-small lung cancer. The US FDA has successfully approved three different categories of immune checkpoint inhibitors (ICIs) such as PD-1 inhibitors (Nivolumab, Pembrolizumab, and Cemiplimab), PDL-1 inhibitors (Atezolimumab, Durvalumab and Avelumab), and CTLA-4 inhibitor (Ipilimumab). Unfortunately, not all patients respond favourably to these drugs, highlighting the role of biomarkers such as Tumour mutation burden (TMB), PDL-1 expression, microbiome, hypoxia, interferon-γ, and ECM in predicting responses to ICIs-based immunotherapy. The current study aims to review the literature and updates on ICIs in cancer therapy.
Collapse
|
7
|
Li Q, Xia L, Sun C, Zhang H, Zheng M, Zhang H, Lu H, Wang Z. Role of Borneol Induced Autophagy in Enhancing Radiosensitivity of Malignant Glioma. Front Oncol 2021; 11:749987. [PMID: 34917504 PMCID: PMC8668811 DOI: 10.3389/fonc.2021.749987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022] Open
Abstract
Glioma is the common primary craniocerebral malignancy with unfavorable prognosis. It is currently treated by surgical resection supplemented by radiotherapy, although the resistance of glioma cells to radiation limits the therapeutic outcomes. The aim of the present study was to determine the potential radiosensitizing effects of borneol and the underlying mechanisms. We found that borneol administration along with radiotherapy significantly inhibited the growth of primary glioma cells in vitro and in vivo. Furthermore, borneol markedly increased the number of autophagosomes in the glioma cells, which coincided with increased expression of beclin-1 and LC3. And the combination of borneol and radiation exposure significantly decreased the expression levels of HIF-1α, mTORC1 and eIF4E. In addition, silencing mTORC1 and eIF4E upregulated Beclin-1 and LC3 and decreased the expression of HIF-1α, thereby inhibiting tumor cell proliferation. Our findings suggest that borneol sensitizes glioma cells to radiation by inducing autophagy via inhibition of the mTORC1/eIF4E/HIF-1α regulatory axis.
Collapse
Affiliation(s)
- Qinglin Li
- Department of Scientific Research, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Liang Xia
- Department of Neurotumor Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Caixing Sun
- Department of Neurotumor Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Huangjie Zhang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Mengying Zheng
- Department of Medical School, Zhejiang University City College, Hangzhou, China
| | - Hongyan Zhang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Hongyang Lu
- Department of Thoracic Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zeng Wang
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
8
|
Wadsworth BJ, Decotret LR, Villamil C, Yapp D, Wilson D, Benard F, McKenzie M, Bennewith KL. Evaluation of 18F-EF5 for detection of hypoxia in localized adenocarcinoma of the prostate. Acta Oncol 2021; 60:1489-1498. [PMID: 34379579 DOI: 10.1080/0284186x.2021.1959636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND A common feature of solid tumours that are resistant to therapy is the presence of regions with low oxygen content (i.e., hypoxia). Oxygen electrode studies suggest that localized prostate adenocarcinoma is commonly hypoxic, although conflicting data have been reported between immunohistochemical detection of hypoxia-induced proteins in biopsy specimens and positron emission tomography (PET) imaging of 18F-labeled hypoxia reporters. Although the 2-nitroimidazole 18F-EF5 is well-established to label hypoxic tumour cells in pre-clinical tumour models and clinical trials of multiple primary tumour sites, it has yet to be tested in prostate cancer. The purpose of this study was to evaluate the feasibility of using 18F-EF5 to detect hypoxia in clinical prostate tumours. MATERIAL AND METHODS Patients with localized adenocarcinoma of the prostate were recruited for pre-treatment 18F-EF5 PET scans. Immunohistochemistry was conducted on diagnostic biopsies to assess the expression of glucose transporter 1 (GLUT1), osteopontin (OPN), and carbonic anhydrase IX (CAIX). Immunoreactivity scores of staining intensity and frequency were used to indicate the presence of tumour hypoxia. RESULTS We found low tumour-to-muscle ratios of 18F-EF5 uptake that were not consistent with tumour hypoxia, causing early termination of the study. However, we observed GLUT1 and OPN expression in all prostate tumour biopsies, indicating the presence of hypoxia in all tumours. CONCLUSION Our data do not support the use of 18F-EF5 PET to detect hypoxia in prostate adenocarcinoma, and suggest the use of immunohistochemistry to quantify expression of the hypoxia-inducible proteins GLUT1 and OPN as indications of prostate tumour hypoxia.
Collapse
Affiliation(s)
- Brennan J. Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Lisa R. Decotret
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Donald Yapp
- Experimental Therapeutics, BC Cancer, Vancouver, Canada
| | - Don Wilson
- Functional Imaging, BC Cancer, Vancouver, Canada
| | - Francois Benard
- Functional Imaging, BC Cancer, Vancouver, Canada
- Molecular Oncology, BC Cancer, Vancouver, Canada
| | | | - Kevin L. Bennewith
- Integrative Oncology, BC Cancer, Vancouver, Canada
- Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
9
|
Mapelli P, Callea M, Fallanca F, Castellano A, Bailo M, Scifo P, Bettinardi V, Conte GM, Monterisi C, Rancoita PMV, Incerti E, Vuozzo M, Gianolli L, Terreni M, Anzalone N, Picchio M. 18F-FAZA PET/CT in pretreatment assessment of hypoxic status in high-grade glioma: correlation with hypoxia immunohistochemical biomarkers. Nucl Med Commun 2021; 42:763-771. [PMID: 33741855 DOI: 10.1097/mnm.0000000000001396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND To investigate the correlation between 18F-labeled fluoroazomycinarabinoside (18F-FAZA) PET data and hypoxia immunohistochemical markers in patients with high-grade glioma (HGG). PATIENTS AND METHODS Prospective study including 20 patients with brain MRI suggestive for HGG and undergoing 18F-FAZA PET/CT before treatment for hypoxia assessment. For each 18F-FAZA PET scan SUVmax, SUVmean and 18F-FAZA tumour volume (FTV) at 40, 50 and 60% threshold of SUVmax were calculated; hypoxic volume was estimated by applying different thresholds (1.2, 1.3 and 1.4) to tumour/blood ratio. Seventeen patients were analysed. The immunohistochemical analysis assessed the following parameters: hypoxia-inducible factor 1α, carbonic anhydrase IX (CA-IX), glucose transporter-1, tumour vascularity and Ki-67. RESULTS 18F-FAZA PET showed a single lesion in 15/17 patients and multiple lesions in 2/17 patients. Twelve/17 patients had grade IV glioma and 5/17 with grade III glioma. Bioptic and surgical samples have been analysed separately. In the surgical subgroup (n = 7) a positive correlation was observed between CA-IX and SUVmax (P = 0.0002), SUVmean40 (P = 0.0058), SUVmean50 (P = 0.009), SUVmean60 (P = 0.0153), FTV-40-50-60 (P = 0.0424) and hypoxic volume1.2-1.3-1.4 (P = 0.0058). In the bioptic group (n = 10) tumour vascularisation was inversely correlated with SUVmax (P = 0.0094), SUVmean40 (P = 0.0107), SUVmean50 (P = 0.0094) and SUVmean60 (P = 0.0154). CONCLUSIONS The correlation of 18F-FAZA PET parameters with CD31 and CA-IX represents a reliable method for assessing tumour hypoxia in HGG. The inverse correlation between tumour vascularisation, SUVmax and SUVmean suggest that highly vascularized tumours might present more oxygen supply than hypoxia.
Collapse
Affiliation(s)
- Paola Mapelli
- Vita-Salute San Raffaele University
- Nuclear Medicine Department
| | | | | | | | - Michele Bailo
- Vita-Salute San Raffaele University
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute
| | | | | | | | | | | | | | | | | | | | | | - Maria Picchio
- Vita-Salute San Raffaele University
- Nuclear Medicine Department
| |
Collapse
|
10
|
Elming PB, Wittenborn TR, Busk M, Sørensen BS, Thomsen MBH, Strandgaard T, Dyrskjøt L, Nielsen S, Horsman MR. Refinement of an Established Procedure and Its Application for Identification of Hypoxia in Prostate Cancer Xenografts. Cancers (Basel) 2021; 13:2602. [PMID: 34073301 PMCID: PMC8198481 DOI: 10.3390/cancers13112602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND This pre-clinical study was designed to refine a dissection method for validating the use of a 15-gene hypoxia classifier, which was previously established for head and neck squamous cell carcinoma (HNSCC) patients, to identify hypoxia in prostate cancer. METHODS PC3 and DU-145 adenocarcinoma cells, in vitro, were gassed with various oxygen concentrations (0-21%) for 24 h, followed by real-time PCR. Xenografts were established in vivo, and the mice were injected with the hypoxic markers [18F]-FAZA and pimonidazole. Subsequently, tumors were excised, frozen, cryo-sectioned, and analyzed using autoradiography ([18F]-FAZA) and immunohistochemistry (pimonidazole); the autoradiograms used as templates for laser capture microdissection of hypoxic and non-hypoxic areas, which were lysed, and real-time PCR was performed. RESULTS In vitro, all 15 genes were increasingly up-regulated as oxygen concentrations decreased. With the xenografts, all 15 genes were up-regulated in the hypoxic compared to non-hypoxic areas for both cell lines, although this effect was greater in the DU-145. CONCLUSIONS We have developed a combined autoradiographic/laser-guided microdissection method with broad applicability. Using this approach on fresh frozen tumor material, thereby minimizing the degree of RNA degradation, we showed that the 15-gene hypoxia gene classifier developed in HNSCC may be applicable for adenocarcinomas such as prostate cancer.
Collapse
Affiliation(s)
- Pernille B. Elming
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Thomas R. Wittenborn
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Morten Busk
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Brita S. Sørensen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
- Danish Center for Particle Therapy, Aarhus University Hospital, 8200 Aarhus, Denmark;
| | - Mathilde Borg Houlberg Thomsen
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus, Denmark; (M.B.H.T.); (T.S.); (L.D.)
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Steffen Nielsen
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| | - Michael R. Horsman
- Experimental Clinical Oncology-Department of Oncology, Aarhus University Hospital, 8200 Aarhus, Denmark; (T.R.W.); (B.S.S.); (S.N.); (M.R.H.)
| |
Collapse
|
11
|
Schüller A, Heinrich S, Fouillade C, Subiel A, De Marzi L, Romano F, Peier P, Trachsel M, Fleta C, Kranzer R, Caresana M, Salvador S, Busold S, Schönfeld A, McEwen M, Gomez F, Solc J, Bailat C, Linhart V, Jakubek J, Pawelke J, Borghesi M, Kapsch RP, Knyziak A, Boso A, Olsovcova V, Kottler C, Poppinga D, Ambrozova I, Schmitzer CS, Rossomme S, Vozenin MC. The European Joint Research Project UHDpulse – Metrology for advanced radiotherapy using particle beams with ultra-high pulse dose rates. Phys Med 2020; 80:134-150. [DOI: 10.1016/j.ejmp.2020.09.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 08/17/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
|
12
|
Beyond tissue biopsy: a diagnostic framework to address tumor heterogeneity in lung cancer. Curr Opin Oncol 2020; 32:68-77. [PMID: 31714259 DOI: 10.1097/cco.0000000000000598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to discuss the strength and limitations of tissue and liquid biopsy and functional imaging to capture spatial and temporal tumor heterogeneity either alone or as part of a diagnostic framework in non-small cell lung cancer (NSCLC). RECENT FINDINGS NSCLC displays genetic and phenotypic heterogeneity - a detailed knowledge of which is crucial to personalize treatment. Tissue biopsy often lacks spatial and temporal resolution. Thus, NSCLC needs to be characterized by complementary diagnostic methods to resolve heterogeneity. Liquid biopsy offers detection of tumor biomarkers and for example, the classification and monitoring of EGFR mutations in NSCLC. It allows repeated sampling, and therefore, appears promising to address temporal aspects of tumor heterogeneity. Functional imaging methods and emerging image analytic tools, such as radiomics capture temporal and spatial heterogeneity. Further standardization of radiomics is required to allow introduction into clinical routine. SUMMARY To augment the potential of precision therapy, improved diagnostic characterization of tumors is pivotal. We suggest a comprehensive diagnostic framework combining tissue and liquid biopsy and functional imaging to address the known aspects of spatial and temporal tumor heterogeneity on the example of NSCLC. We envision how this framework might be implemented in clinical practice.
Collapse
|
13
|
Waller J, Onderdonk B, Flood A, Swartz H, Shah J, Shah A, Aydogan B, Halpern H, Hasan Y. The clinical utility of imaging methods used to measure hypoxia in cervical cancer. Br J Radiol 2020; 93:20190640. [PMID: 32286849 PMCID: PMC7336054 DOI: 10.1259/bjr.20190640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 03/18/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
While it is well-established that hypoxia is a major factor that affects clinical outcomes in cervical cancer, widespread usage of clinically available methods to detect and evaluate hypoxia during the course of treatment have not been established. This review compares these methods, summarizes their strengths and weaknesses, and assesses the pathways for their useful employment to alter clinical practice. We conducted a search on PubMed for literature pertaining to imaging hypoxic cervical cancer, and implemented keywords related to oxygen measurement tools to improve the relevance of the search results.Oxygenation level-dependent applications of MRI have demonstrated hypoxia-induced radioresistance, and changes in cervix tumor oxygenation from hyperoxic therapy.The hypoxic areas within tumors can be indirectly identified in dynamic contrast-enhanced images, where they generally display low signal enhancement, and diffusion-weighted images, which demonstrates areas of restricted diffusion (which correlates with hypoxia). Positron emmision tomography, used independently and with other imaging modalities, has demonstrated utility in imaging hypoxia through tracers specific for low oxygen levels, like Cu-ATSM tracers and nitroimidazoles. Detecting hypoxia in the tumors of patients diagnosed with cervical cancer via medical imaging and non-imaging tools like electron paramagnetic resonance oximetry can be utilized clinically, such as for guiding radiation and post-treatment surveillance, for a more personalized approach to treatment. The merits of these methods warrant further investigation via comparative effectiveness research and large clinical trials into their clinical applications.
Collapse
Affiliation(s)
- Joseph Waller
- Drexel College of Medicine, 2900 W Queen Ln, PA 19129, United States
| | - Benjamin Onderdonk
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Ann Flood
- Department of Radiology, Dartmouth Geisel School of Medicine, 1 Rope Ferry Rd, NH 03755, United States
| | - Harold Swartz
- Department of Radiology, Dartmouth Geisel School of Medicine, 1 Rope Ferry Rd, NH 03755, United States
| | - Jaffer Shah
- Drexel College of Medicine, 2900 W Queen Ln, PA 19129, United States
| | - Asghar Shah
- Brown University, Providence, RI 02912, United States
| | - Bulent Aydogan
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Howard Halpern
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| | - Yasmin Hasan
- Department of Radiation and Cellular Oncology, The University of Chicago, 5758 S Maryland Ave, IL 60637, United States
| |
Collapse
|
14
|
Clinical and Statistical Considerations when Assessing Oxygen Levels in Tumors: Illustrative Results from Clinical EPR Oximetry Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1232:155-168. [PMID: 31893406 DOI: 10.1007/978-3-030-34461-0_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The success of treatment for malignancies, especially those undergoing radiation therapy or chemotherapy, has long been recognized to depend on the degree of hypoxia in the tumor. In addition to the prognostic value of knowing the tumor's initial level of hypoxia, assessing the tumor oxygenation during standard therapy or oxygen-related treatments (such as breathing oxygen-enriched gas mixtures or taking drugs that can increase oxygen supply to tissues) can provide valuable data to improve the efficacy of treatments. A series of early clinical studies of tumors in humans are ongoing at Dartmouth and Emory using electron paramagnetic resonance (EPR) oximetry to assess tumor oxygenation, initially and over time during either natural disease progression or treatment. This approach has the potential for reaching the long-sought goal of enhancing the effectiveness of cancer therapy. In order to effectively reach this goal, we consider the validity of the practical and statistical assumptions when interpreting the measurements made in vivo for patients undergoing treatment for cancer.
Collapse
|
15
|
Pratx G, Kapp DS. A computational model of radiolytic oxygen depletion during FLASH irradiation and its effect on the oxygen enhancement ratio. Phys Med Biol 2019; 64:185005. [PMID: 31365907 DOI: 10.1088/1361-6560/ab3769] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recent results from animal irradiation studies have demonstrated the potential of ultra-high dose rate irradiation (also known as FLASH) for reducing radiation toxicity in normal tissues. However, despite mounting evidence of a 'FLASH effect', a mechanism has yet to be elucidated. This article hypothesizes that the radioprotecting effect of FLASH irradiation could be due to the specific sparing of hypoxic stem cell niches, which have been identified in several organs including the bone marrow and the brain. To explore this hypothesis, a new computational model is presented that frames transient radiolytic oxygen depletion (ROD) during FLASH irradiation in terms of its effect on the oxygen enhancement ratio (OER). The model takes into consideration oxygen diffusion through the tissue, its consumption by metabolic cells, and its radiolytic depletion to estimate the relative decrease in radiosensitivity of cells receiving FLASH irradiation. Based on this model and the following parameters (oxygen diffusion constant [Formula: see text] = 2 · 10-5 cm2 s-1, oxygen metabolic rate m = 3 mmHg s-1, ROD rate L ROD = [Formula: see text] mmHg Gy-1, prescribed dose D p = 10 Gy, and capillary oxygen tension p 0 = 40 mmHg), several predictions are made that could be tested in future experiments: (1) the FLASH effect should gradually disappear as the radiation pulse duration is increased from <1 s to 10 s; (2) dose should be deposited using the smallest number of radiation pulses to achieve the greatest FLASH effect; (3) a FLASH effect should only be observed in cells that are already hypoxic at the time of irradiation; and (4) changes in capillary oxygen tension (increase or decrease) should diminish the FLASH effect.
Collapse
Affiliation(s)
- Guillem Pratx
- 300 Pasteur Dr, Grant S277, Stanford, CA 94305-5132, United States of America. Author to whom any correspondence should be addressed
| | | |
Collapse
|
16
|
Daimiel I. Insights into Hypoxia: Non-invasive Assessment through Imaging Modalities and Its Application in Breast Cancer. J Breast Cancer 2019; 22:155-171. [PMID: 31281720 PMCID: PMC6597408 DOI: 10.4048/jbc.2019.22.e26] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
Oxygen is crucial to maintain the homeostasis in aerobic cells. Hypoxia is a condition in which cells are deprived of the oxygen supply necessary for their optimum performance. Whereas oxygen deprivation may occur in normal physiological processes, hypoxia is frequently associated with pathological conditions. It has been identified as a stressor in the tumor microenvironment, acting as a key mediator of cancer development. Numerous pathways are activated in hypoxic cells that affect cell signaling and gene regulation to promote the survival of these cells by stimulating angiogenesis, switching cellular metabolism, slowing their growth rate, and preventing apoptosis. The induction of dysregulated metabolism in cancer cells by hypoxia results in aggressive tumor phenotypes that are characterized by rapid progression, treatment resistance, and poor prognosis. A non-invasive assessment of hypoxia-induced metabolic and architectural changes in tumors is advisable to fully improve breast cancer (BC) patient management, by potentially reducing the need for invasive biopsy procedures and evaluating tumor response to treatment. This review provides a comprehensive overview of the molecular changes in breast tumors secondary to hypoxia and the non-invasive imaging alternatives to evaluate oxygen deprivation, with an emphasis on their application in BC and the advantages and limitations of the currently available techniques.
Collapse
Affiliation(s)
- Isaac Daimiel
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
17
|
Advanced PET imaging in oncology: status and developments with current and future relevance to lung cancer care. Curr Opin Oncol 2019; 30:77-83. [PMID: 29251666 DOI: 10.1097/cco.0000000000000430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW This review highlights the status and developments of PET imaging in oncology, with particular emphasis on lung cancer. We discuss the significance of PET for diagnosis, staging, decision-making, monitoring of treatment response, and drug development. The PET key advantage, the noninvasive assessment of functional and molecular tumor characteristics including tumor heterogeneity, as well as PET trends relevant to cancer care are exemplified. RECENT FINDINGS Advances of PET and radiotracer technology are encouraging for multiple fields of oncological research and clinical application, including in-depth assessment of PET images by texture analysis (radiomics). Whole body PET imaging and novel PET tracers allow assessing characteristics of most types of cancer. However, only few PET tracers in addition to F-fluorodeoxyglucose have sufficiently been validated, approved, and are reimbursed for a limited number of indications. Therefore, validation and standardization of PET parameters including tracer dosage, image acquisition, post processing, and reading are required to expand PET imaging as clinically applicable approach. SUMMARY Considering the potential of PET imaging for precision medicine and drug development in lung and other types of cancer, increasing efforts are warranted to standardize PET technology and to provide evidence for PET imaging as a guiding biomarker in nearly all areas of cancer treatment.
Collapse
|
18
|
2-Nitroimidazole-Furanoside Derivatives for Hypoxia Imaging-Investigation of Nucleoside Transporter Interaction, 18F-Labeling and Preclinical PET Imaging. Pharmaceuticals (Basel) 2019; 12:ph12010031. [PMID: 30781409 PMCID: PMC6469291 DOI: 10.3390/ph12010031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/04/2019] [Accepted: 02/12/2019] [Indexed: 11/16/2022] Open
Abstract
The benefits of PET imaging of tumor hypoxia in patient management has been demonstrated in many examples and with various tracers over the last years. Although, the optimal hypoxia imaging agent has yet to be found, 2-nitroimidazole (azomycin) sugar derivatives—mimicking nucleosides—have proven their potential with [18F]FAZA ([18F]fluoro-azomycin-α-arabinoside) as a prominent representative in clinical use. Still, for all of these tracers, cellular uptake by passive diffusion is postulated with the disadvantage of slow kinetics and low tumor-to-background ratios. We recently evaluated [18F]fluoro-azomycin-β-deoxyriboside (β-[18F]FAZDR), with a structure more similar to nucleosides than [18F]FAZA and possible interaction with nucleoside transporters. For a deeper insight, we comparatively studied the interaction of FAZA, β-FAZA, α-FAZDR and β-FAZDR with nucleoside transporters (SLC29A1/2 and SLC28A1/2/3) in vitro, showing variable interactions of the compounds. The highest interactions being for β-FAZDR (IC50 124 ± 33 µM for SLC28A3), but also for FAZA with the non-nucleosidic α-configuration, the interactions were remarkable (290 ± 44 µM {SLC28A1}; 640 ± 10 µM {SLC28A2}). An improved synthesis was developed for β-FAZA. For a PET study in tumor-bearing mice, α-[18F]FAZDR was synthesized (radiochemical yield: 15.9 ± 9.0% (n = 3), max. 10.3 GBq, molar activity > 50 GBq/µmol) and compared to β-[18F]FAZDR and [18F]FMISO, the hypoxia imaging gold standard. We observed highest tumor-to-muscle ratios (TMR) for β-[18F]FAZDR already at 1 h p.i. (2.52 ± 0.94, n = 4) in comparison to [18F]FMISO (1.37 ± 0.11, n = 5) and α-[18F]FAZDR (1.93 ± 0.39, n = 4), with possible mediation by the involvement of nucleoside transporters. After 3 h p.i., TMR were not significantly different for all 3 tracers (2.5–3.0). Highest clearance from tumor tissue was observed for β-[18F]FAZDR (56.6 ± 6.8%, 2 h p.i.), followed by α-[18F]FAZDR (34.2 ± 7.5%) and [18F]FMISO (11.8 ± 6.5%). In conclusion, both isomers of [18F]FAZDR showed their potential as PET hypoxia tracers. Differences in uptake behavior may be attributed to a potential variable involvement of transport mechanisms.
Collapse
|
19
|
You GR, Cheng AJ, Lee LY, Huang YC, Liu H, Chen YJ, Chang JT. Prognostic signature associated with radioresistance in head and neck cancer via transcriptomic and bioinformatic analyses. BMC Cancer 2019; 19:64. [PMID: 30642292 PMCID: PMC6332600 DOI: 10.1186/s12885-018-5243-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/21/2018] [Indexed: 02/08/2023] Open
Abstract
Background Radiotherapy is an indispensable treatment modality in head and neck cancer (HNC), while radioresistance is the major cause of treatment failure. The aim of this study is to identify a prognostic molecular signature associated with radio-resistance in HNC for further clinical applications. Methods Affymetrix cDNA microarrays were used to globally survey different transcriptomes between HNC cell lines and isogenic radioresistant sublines. The KEGG and Partek bioinformatic analytical methods were used to assess functional pathways associated with radioresistance. The SurvExpress web tool was applied to study the clinical association between gene expression profiles and patient survival using The Cancer Genome Atlas (TCGA)-head and neck squamous cell carcinoma (HNSCC) dataset (n = 283). The Kaplan-Meier survival analyses were further validated after retrieving clinical data from the TCGA-HNSCC dataset (n = 502) via the Genomic Data Commons (GDC)-Data-Portal of National Cancer Institute. A panel maker molecule was generated to assess the efficacy of prognostic prediction for radiotherapy in HNC patients. Results In total, the expression of 255 molecules was found to be significantly altered in the radioresistant cell sublines, with 155 molecules up-regulated 100 down-regulated. Four core functional pathways were identified to enrich the up-regulated genes and were significantly associated with a worse prognosis in HNC patients, as the modulation of cellular focal adhesion, the PI3K-Akt signaling pathway, the HIF-1 signaling pathway, and the regulation of stem cell pluripotency. Total of 16 up-regulated genes in the 4 core pathways were defined, and 11 over-expressed molecules showed correlated with poor survival (TCGA-HNSCC dataset, n = 283). Among these, 4 molecules were independently validated as key molecules associated with poor survival in HNC patients receiving radiotherapy (TCGA-HNSCC dataset, n = 502), as IGF1R (p = 0.0454, HR = 1.43), LAMC2 (p = 0.0235, HR = 1.50), ITGB1 (p = 0.0336, HR = 1.46), and IL-6 (p = 0.0033, HR = 1.68). Furthermore, the combined use of these 4 markers product an excellent result to predict worse radiotherapeutic outcome in HNC (p < 0.0001, HR = 2.44). Conclusions Four core functional pathways and 4 key molecular markers significantly contributed to radioresistance in HNC. These molecular signatures may be used as a predictive biomarker panel, which can be further applied in personalized radiotherapy or as radio-sensitizing targets to treat refractory HNC. Electronic supplementary material The online version of this article (10.1186/s12885-018-5243-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guo-Rung You
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ann-Joy Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Yu-Chen Huang
- Department of Oral Maxillofacial Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Hsuan Liu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Joseph T Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan. .,Department of Radiation Oncology, Xiamen Chang Gung Memorial Hospital, Xiamen, Fujian, China.
| |
Collapse
|
20
|
Crispin-Ortuzar M, Apte A, Grkovski M, Oh JH, Lee NY, Schöder H, Humm JL, Deasy JO. Predicting hypoxia status using a combination of contrast-enhanced computed tomography and [ 18F]-Fluorodeoxyglucose positron emission tomography radiomics features. Radiother Oncol 2018; 127:36-42. [PMID: 29273260 PMCID: PMC5924729 DOI: 10.1016/j.radonc.2017.11.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/11/2017] [Accepted: 11/26/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE Hypoxia is a known prognostic factor in head and neck cancer. Hypoxia imaging PET radiotracers such as 18F-FMISO are promising but not widely available. The aim of this study was therefore to design a surrogate for 18F-FMISO TBRmax based on 18F-FDG PET and contrast-enhanced CT radiomics features, and to study its performance in the context of hypoxia-based patient stratification. METHODS 121 lesions from 75 head and neck cancer patients were used in the analysis. Patients received pre-treatment 18F-FDG and 18F-FMISO PET/CT scans. 79 lesions were used to train a cross-validated LASSO regression model based on radiomics features, while the remaining 42 were held out as an internal test subset. RESULTS In the training subset, the highest AUC (0.873±0.008) was obtained from a signature combining CT and 18F-FDG PET features. The best performance on the unseen test subset was also obtained from the combined signature, with an AUC of 0.833, while the model based on the 90th percentile of 18F-FDG uptake had a test AUC of 0.756. CONCLUSION A radiomics signature built from 18F-FDG PET and contrast-enhanced CT features correlates with 18F-FMISO TBRmax in head and neck cancer patients, providing significantly better performance with respect to models based on 18F-FDG PET only. Such a biomarker could potentially be useful to personalize head and neck cancer treatment at centers for which dedicated hypoxia imaging PET radiotracers are unavailable.
Collapse
Affiliation(s)
- Mireia Crispin-Ortuzar
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA; Cancer Research UK Cambridge Institute, University of Cambridge, UK.
| | - Aditya Apte
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
21
|
Jiang X, Zhang B, Zhou Z, Meng L, Sun Z, Xu Y, Xu Q, Yuan A, Yu L, Qian H, Wu J, Hu Y, Liu B. Enhancement of radiotherapy efficacy by pleiotropic liposomes encapsulated paclitaxel and perfluorotributylamine. Drug Deliv 2017; 24:1419-1428. [PMID: 28937321 PMCID: PMC8241066 DOI: 10.1080/10717544.2017.1378939] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 09/09/2017] [Accepted: 09/09/2017] [Indexed: 01/05/2023] Open
Abstract
Paclitaxel (PTX) is widely used as a radiosensitizer in the clinical treatment of cancer. However, the efficacy of chemoradiotherapy is limited by the hostility of the tumor microenvironment such as hypoxia. To overcome this constraint, we designed pleiotropic radiotherapy sensitized liposomes containing perfluorotributylamine (PFTBA) and PTX. The results showed that liposomes significantly accumulated in the tumor site. PFTBA in liposomes dramatically reversed tumor hypoxia and improved the sensitivity of tumor radiotherapy. PTX in liposomes blocked the cell cycle of tumor cells in the radiation-sensitive G2/M phase, which was even greater when combined with PFTBA. In vitro and in vivo tumor treatment further demonstrated remarkably improved therapeutic outcomes in radiotherapy with such biocompatible liposomes. In conclusion, the pleiotropic liposomes encapsulated PFTBA and PTX provide significant radiotherapy sensitization and show promise for future application in clinical medicine.
Collapse
Affiliation(s)
- Xing Jiang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- College of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Baoli Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Zaigang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Lingtong Meng
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Zhiling Sun
- College of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Xu
- College of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiuping Xu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
22
|
Wadsworth BJ, Pan J, Dude I, Colpo N, Bosiljcic M, Lin KS, Benard F, Bennewith KL. 2-18F-Fluoroethanol Is a PET Reporter of Solid Tumor Perfusion. J Nucl Med 2017; 58:815-820. [DOI: 10.2967/jnumed.116.183624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/21/2016] [Indexed: 11/16/2022] Open
|
23
|
Colliez F, Gallez B, Jordan BF. Assessing Tumor Oxygenation for Predicting Outcome in Radiation Oncology: A Review of Studies Correlating Tumor Hypoxic Status and Outcome in the Preclinical and Clinical Settings. Front Oncol 2017; 7:10. [PMID: 28180110 PMCID: PMC5263142 DOI: 10.3389/fonc.2017.00010] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/10/2017] [Indexed: 12/30/2022] Open
Abstract
Tumor hypoxia is recognized as a limiting factor for the efficacy of radiotherapy, because it enhances tumor radioresistance. It is strongly suggested that assessing tumor oxygenation could help to predict the outcome of cancer patients undergoing radiation therapy. Strategies have also been developed to alleviate tumor hypoxia in order to radiosensitize tumors. In addition, oxygen mapping is critically needed for intensity modulated radiation therapy (IMRT), in which the most hypoxic regions require higher radiation doses and the most oxygenated regions require lower radiation doses. However, the assessment of tumor oxygenation is not yet included in day-to-day clinical practice. This is due to the lack of a method for the quantitative and non-invasive mapping of tumor oxygenation. To fully integrate tumor hypoxia parameters into effective improvements of the individually tailored radiation therapy protocols in cancer patients, methods allowing non-invasively repeated, safe, and robust mapping of changes in tissue oxygenation are required. In this review, non-invasive methods dedicated to assessing tumor oxygenation with the ultimate goal of predicting outcome in radiation oncology are presented, including positron emission tomography used with nitroimidazole tracers, magnetic resonance methods using endogenous contrasts (R1 and R2*-based methods), and electron paramagnetic resonance oximetry; the goal is to highlight results of studies establishing correlations between tumor hypoxic status and patients’ outcome in the preclinical and clinical settings.
Collapse
Affiliation(s)
- Florence Colliez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| |
Collapse
|